EXAM #2: ANTINEOPLASTIC AGENTS Flashcards

1
Q

What are the three major cell cycle checkpoints?

A

G1/S
G2/M
Metaphase/Anaphase

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

What are the specific criteria necessary to proceed through the G1/S checkpoint?

A

1) Cell nutrition, size, and environment must be favorable for replication
2) DNA must be intact

This check ensures the cell is prepared for DNA replication and to enter S-phase.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What are the specific criteria necessary to proceed through the G2/M checkpoint?

A

DNA must be completely replicated

This is the checkpoint that ensures the cell is ready to enter mitosis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What are the specific criteria necessary to proceed through the metaphase/anaphase checkpoint?

A

1) DNA intact must be intact
2) Chromosomes must be attached to the mitotic spindle

This check ensure that the cell is ready for chromatid separation and is prepared for cytokinesis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What are the three mechanisms of oncogene formation?

A

First, an oncogene is a mutated form of a normal gene that supported cellular proliferation i.e. “proto-oncogenes”

1) Point mutations
2) Chromosomal translocation
3) Proto-oncogene duplication/ amplification

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

How can point mutations lead to the development of an oncogene?

A
  • Change in an amino acid regulatory region in the proto-oncogene product
  • Amino acid change that makes the proto-oncogene resistant to degradation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

How do chromosomal translocations result in oncogenes? Give an example.

A

Chromosomal translocation that causes the fusion of two genes, the product of which is the “fusion gene”

E.g. “Philadelphia” chromosome (9 x 22) ABL and BCR in CML

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

What is a major oncogene that is found in roughly 30% of all cancers?

A

Ras

Ras in involved in cell signaling/ GPCR signaling; mutations leave Ras permanently turned on.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Define first order killing of cancer cells i.e. what is the cell kill hypothesis?

A

This is the chemotheraputic elimination of a FIXED percentage of cancer cells.

This is first order kinetics*

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What are the four goals of cancer drug therapy?

A

1) Curative intent
2) Adjuvant therapy
3) Neoadjuvant therapy
4) Palliation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

List the general characteristics of chemotheraputic drug therapy.

A
  • Drugs are cytotoxic and only PARTIALLY selective
  • Low TI

Thus, adverse effects are common.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

What are the general mechanisms of resistance to chemotherapeutic agents?

A

1) Genetic instability
2) Drug efflux pumps/ mutated drug transporters
3) DNA damage repair
4) Cell death inhibition
5) Drug inactivation
- Up-regulation of enzymes that metabolize drugs
6) Drug target alteration
7) Epithelial-Mesenchymal Transition

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What types of tumors are easiest to treat with chemotherapy?

A

Tumors that are RAPIDLY growing

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Define curative intent.

A

Drug therapy is intended to CURE the disease

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Define adjuvant therapy.

A
  • Drug given AFTER primary treatment e.g. surgery

- Prevent recurrence

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Define neoadjuvant therapy.

A

Drug given FIRST to SHRINK tumor, making it amenable to surgery

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

Define palliative therapy.

A

Therapy intended to relieve symptoms and improve quality of life.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Do most cancers follow the cell-kill hypothesis? Why or why not?

A

NO

B/c of the cytotoxicity of chemotherapeutic, it is not possible to dose patients in a manner necessary to achieve 1st order kill.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

What is a drug efflux pump?

A

A cellular transporter than “pumps” drugs out

E.g. MDR-2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

What is Epithelial- Mesenchymal Transition (EMT)?

A
  • Cell loses adhesive properties and becomes motile

- Changes can also cause drug resistance

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

What are the three general cytotoxic mechanisms to kill cancer cells?

A

1) Perturb normal DNA replication
2) Perturb mitosis
3) Starve cells of amino acids

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What are the three targeted mechanisms to kill cancer cells?

A

1) Perturb hormone and growth factor signaling
2) Inhibit blood supply to tumor e.g. VEGF
3) Target activating proteins

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What are cell-cycle non-specific drugs?

A

DNA alkylating agents that kill cells in ANY stage

Preferentially kill replicating cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

What are the S-phase specific drugs?

A

DNA synthesis inhibitors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

What is the mechanism of action of the “antimetabolites?”

A

Inhibiting de novo nucleotide biosynthesis and generally target cells in S-phase

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

What is the function of ribonucleotide reductase?

A

Turning ribonucleotides into deoxyribnucleotides

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

What is the mechanism of action of methotrexate?

A
  • Anti-metabolite that targets s-phase
  • Inhibition of dihydrofolate reductase i.e. the enyzme that makes THF from dihydrofolate
  • THF is crucial in the action of thymidylate synthetase (dUMP–>dTMP), which is required for DNA synthesis

Essentially, inhibiting DNA synthesis by preventing the action of folate

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

Why can extremely high does of Methotrexate be administered to pateints?

A

Leucovorin rescue (folinic acid)

Leucovorin is a reduced form of folic acid. Administration within the proper window can allow for nearly fatal doses of methotrexate do be administered (hopefully targeting cancerous/ rapidly dividing cell), followed by Leucovorin rescue to prevent harmful toxic effects of such a high dose to normal tissue.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

What are the mechanisms of Methotrexate resistance?

A

1) Impaired transport
2) Altered DHFR (isozyme) that decrease the affinity of methotrexate’s target
3) Elevated DHFR expression

DHFR= dihydrofolate reductase

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

What are the unique toxicities seen with Methotrexate?

A
  • Interstitial pneumonitis

- Nephrotoxicity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

What is the mechanism of action of 5-fluorouracil?

A
  • Pyrimidine analog
  • Anti-metabolite that targets s-phase
  • 5-FU is metabolized:
    1) 5dUMP, which INHIBITS THYMIDYLATE SYNTHASE and prevents DNA synthesis
    2) FUTP that is falsely incorporated into RNA
    3) FdUTP that is falsely incorporated into DNA

False incorporation causes defects in RNA/DNA function and sturucture leading to cell death.*

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

What are the unique adverse effects seen with 5-FU administration?

A

Oral and GI ulcers

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

What is Capecitabine?

A
  • Antimetabolite
  • Targets S-phase

This Prodrug of 5-FU that can be given orally; same mechanism of action as 5-FU (inhibition of thymidylate synthase etc.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

What is Cytarabine used to treat?

A

This is the most important antimetabolite for AML

It is only used to treat HEMATOLOGIC MALIGNANCIES*

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

What is the mechanism of action of Cytarabine?

A
  • Pyrimidine analog/ anti-metabolite
  • Targets cells in S-phase

1) Cytarabine or Ara-C is transported into the cell and converted to Ara-CMP by DEOXYCYTIDINE KINASE
2) Ara-CMP is converted to Ara-dCTP
3) Ara-dCTP is incorporated into DNA and inhibits DNA synthesis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

What is the specific toxicity that is associated with Cytarabine?

A

Cerebellar Syndrome

*****Note that inactivation of Ara-C required CYTIDINE DEAMINASE; there are low levels of this enzyme in the brain, making brain more susceptible to adverse effects of drug.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

What are the symptoms of Cerebellar Syndrome?

A

1) Dysarthria
2) Nystagumus
3) Ataxia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

What are the major mechanisms of cytarabine resistance?

A

1) Loss of DEOXYCYTIDINE KINASE
2) Inability of tumor cells to transport Ara-C into cells
3) Cytidine demainase upregulation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

What is the mechanism of action of Gemcitabine?

A
  • Pyrimidine analog/ anti-metabolite that targets S-phase

1) Converted to active form by DEOXYCYTIDINE KINASE, like Cytarabine
1) Incorporated into DNA–inhibits DNA synthesis
3) Inhibits RIBONUCLEOTIDE REDUCTASE, which also inhibits DNA synthesis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

What are the mechanisms of Gemcitabine resistance?

A

1) Reduced activity of DEOXYCYTIDINE KINASE

2) Increased production of deoxycytidine b/c of inhibitive effect on deoxycytidine kinase

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

What is the mechanism of action of 6-MP and 6-TG?

A
  • Purine analogs/ antimetabolites that inhibit s-phase of the cell cycle

1) Activated by HGPRT into thio-IMP and thio-GMP respectively
2) thio-IMP converted into thio-GMP
3) thio-GMP blocks BOTH SALVAGE and DE NOVO purine synthesis i.e. inhibits DNA replication

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

What is the common mechanism of resistance in 6-MP and 6-TG?

A

Decreased HGPRT activity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

What enzymes inactivates 6-MP? Why is this important?

A

Thiopurine methyltransferase (TPMT)

Polymorphism causes reduced TPMT activity that can lead to life-threatening toxicity and need for lower doses

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

What is the mechanism of action of Fludarabine?

A
  • Purine analog/ antimetabolite that targets cells in s-phase

1) Activated by DEOXYCYTIDINE KINASE and incorporated into DNA and RNA
2) Inhibits RIBONUCLEOTIDE REDUCTASE and DNA POLYMERASE
3 Inhibits RNA function and mRNA translation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

What is the common mechanism of Fludarabine resistance?

A

Decreased of DEOXYCYTIDINE KINASE

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

What is the mechanism of action of Cladribine?

A
  • Purine analog/ antimetabolite that targets cells in s-phase

1) Activated by DEOXYCYTIDINE KINASE and incorporated into DNA
2) Causes DNA strand breaks
3) Inhibitor of RIBONUCLEOTIDE REDUCTASE

47
Q

List the alkylating agents.

A

Nitrogen mustards

  • Mechlorethamine
  • Cyclophosphamide

Nitrosureas
- Carmustine (BCNU)

48
Q

What is the general mechanism of action of the alkylating agents? What phase of the cell cycle do they target?

A
  • Alkylation of the N7 position on guanine to cause DNA cross-linking and strand breakage
  • Cytotoxic to the WHOLE cell cycle
49
Q

What is the mechanism of action of Cyclophosmphamine?

A
  • Prodrug that is metabolized to active form
  • Alkylating agent

Note that the active drug can be converted to another drug, “Acrolein” which is specifically cytotoxic to the bladder

50
Q

What are the specific toxicities associated with Cyclophosphamide?

A

Hemorrhagic cystitis

Recall, it is a prodrug that is converted to an active form; active form is converted to ACROLEIN, which is specifically toxic to the bladder.

51
Q

What drug can be administered with Cyclophosphamine to reduce the risk of hemorrhagic cystitis?

A

Mensa

This drug INACTIVATES ACROLEIN

52
Q

What is Carmustine (BCNU) commonly used to treat? Why?

A
  • Alkylating agent
  • Used to treat BRAIN TUMORS b/c it is highly lipophilic

Often included in wafers that are placed post-op*

53
Q

List the general adverse effects of alkylating agents.

A

1) Mutagenic, teratogenic, myelosuppressive
2) Dose-limiting factor is bone marrow suppression and damage to intestinal mucosa
3) Cause Leukemia
4) Strong vesicant/ blistering properties

54
Q

What are the mechanisms of resistance to the alkylating agents?

A
  • Inactivation by GLUTATHIONE/ increased production of
  • Reduced uptake
  • Accelerated DNA repair/ double strand break repair
  • Increased expression of MGMT, which removes alkyl groups from guanine before cross-links form
55
Q

What is the function of MGMT?

A

Prevention of DNA damage by removing alkly groups from guanine i.e. prevents drugs from “alkylating”

56
Q

List the non-classical alkylating agents.

A

Platinum compounds

  • Cisplatin
  • Carboplatin
  • Oxaliplatin
57
Q

What is the mechanism of action of the platinum compounds?

A
  • Generally, these drugs cause DNA cross-linking WITHOUT actually alkylating DNA
  • Active throughout the whole cell cycle

Note that these compounds are actively transported into the cell via the Cu++ transporter

58
Q

What are the side effects that are specific to Cisplatin?

A
  • Neurotoxicity i.e. peripheral motor and sensory neuropathy
  • Nephrotoxicity
  • Anaphylaxis
59
Q

What are the side effects that are specific to Carboplatin?

A

Less nausea, neuropathy and nephrotoxicity, but can induce anaphylaxis like Cisplatin

60
Q

How is the nephrotoxicity of Cisplatin decreased?

A

Coadministration of IV saline

61
Q

What is Procarbazine used to treat?

A

This is a non-classical alkylating agent used to treat Hodgkin’s Disease

62
Q

What is Dacarbazine used to treat?

A

This is a non-classical alkylating agent used to treat Hodgkin’s lymphoma, melanoma, and sarcoma

63
Q

What is Temozolamide used to treat?

A

This is a non-classical alkylating agent used to treat Glioblastomas and metastatic melanoma

64
Q

Describe microtubule dynamic instability.

A
  • Equilibrium between forming and falling apart

- MTs need to be able to do BOTH function normally

65
Q

List the antimicrotubule agents.

A

Vinblastine
Vincristine
Paclitaxel
Docetaxel

66
Q

What is the mechanism of action of Vinblastin and Vinctistine? What cell cycle phase do they kill in?

A
  • Vinblastine and Vincristine prevent MT polymeratiztion

- Without properly forming MTs, the mitotic spindle cannot form; thus, these compounds kill in MITOSIS

67
Q

What is the major adverse effect of vincristine?

A

Neurotoxicity

Note that this is NOT seen with Vinblastine but does have myelosuppression*

68
Q

What is the mechanism of action of the Taxanes? What cell cycle phase do they kill in?

A
  • Prevent MT destruction/ depolymerization

- Thus, the mitotic spindle can form in MITOSIS, but the PULLING phase is inhibited

69
Q

What is the major adverse effect of Paclitaxel?

A

1) Peripheral neuropathy
2) Mylosuppression
3) Anaphylaxis in ~5% of patients

70
Q

What drugs are commonly given with Paclitaxel?

A

1) Filgrastim= G-CSF to reduce myelosuppression

2) Dexamethasone and antihistamines that prevent hypersensitivity reactions

71
Q

What is the normal function of Topoisomersase in the cell?

A
  • Recall that DNA is normally stored in a condensed form

- TOPOISOMERASE cuts DNA for replication, unwinds it, and repairs the cut

72
Q

List the Topoisomerase I inhibitors.

A

Irinotecan

Topotecan

73
Q

List the Topoisomerase II inhibitors.

A

Etoposide

74
Q

What is the mechanism of action of Doxorubicin?

A

Anthracycline antibiotic

  • Intercalates with DNA and inhibits DNA polymerase
  • Inhibits topoisomerase II
75
Q

What is the major adverse effect seen with Doxorubicin?

A
  • Binds iron and generates ROS

- Causes irreversible CARDIOMYPOATHY

76
Q

What can reduce the cardiotoxicity of Doxorubicin?

A

Dexrazoxane

This is an iron chelator that reduces the formation of free radical formation

77
Q

What is the mechanism of action of Bleomycin?

A

This is a small peptide that binds DNA and causes single and double strand breaks

78
Q

What phase of the cell cycle does Bleomycin target?

A

G2

79
Q

What is unique about the toxicity of Bleomycin?

A

Cumulative, irreversible PULMONARY toxicity

Only minimally myelosuppressive*

80
Q

What is the clincal utility of Glucocorticoids in cancer treatment?

A

1) Inhibit lymphocyte proliferation
2) Reduce ICP w/ brain tumors
3) Reduce nausea and vomiting associated with chemotherapy

81
Q

What is the mechanism of action of Tamoxifen?

A

Partial estrogen receptor antagonist

82
Q

What cancer is Tamoxifen used to treat?

A

Breast cancer

83
Q

How is estrogen synthesized in post-menopausal women?

A

In post-menopausal women, AROMATASE converts testosterone into estrogen in peripheral tissues

84
Q

What is the mechanism of action of Anastrazole? What cancer is it used to treat?

A

Anaztrazole is an aromatase antagonist used to treat breast cancer that is estrogen receptor positive in post-menopausal women

85
Q

What are androgen receptor inhibitors used to treat? What is the mechanism of action?

A

Prostate cancer

These drugs prevent DHT from binding to androgen receptors, which normally drives prostate cancer growth

86
Q

What are Leuprolide and goserelin?

A
  • GnRH AGONISTS
  • Causes DESENSITIZATION of the GnRH receptor in the pituitary in the long-term
  • Decreases LH and FSH
  • Decreases Testosterone

Used to treat prostate cancer*

87
Q

What is the mechanism of Degarelix?

A

GnRH ANTAGONIST used to treat prostate cancer

88
Q

What is the mechanism of action of Trastuzumab?

A

Monoclonal antibody that targets HER-2 (EGFR TRK amplified in invasive breast cancer) and prevents signaling

Used to treat HER-2+ breast cancer

89
Q

What is the major toxicity associated with Trastuzumab?

A

Cardiotoxicity

90
Q

What is the mechanism of Cetuximab?

A

Monocolonal antibody used to treat EGFR+ COLORECTAL tumors

91
Q

What is a mechanism of Cetuximab resistance?

A

Activating mutations in Ras

Thus, Ras activity is tested prior to use of this drug*

92
Q

What is Bevacizumb’s mechanism of action?

A

Monoclonal antibody for VEGF i.e. prevent angiogenesis

93
Q

What are the adverse effects assocaited with Bevacizumb?

A
  • HTN
  • Increased risk of bleeding/ thrombosis
  • Increased risk of GI perforation
  • Decreased wound healing
94
Q

What is the mechanism of action of Lapatinib?

A

Small molecule that inhibits EGFR and HER-2 kinase activity in THE CELL

Used specifically with capecitabine to treat HER-2+, Trastuzumab refractory breast cancer*

95
Q

What is the mechanism of action of Erlotinib?

A

Small molecule EGFR inhibitor as a first line treatment for NON-SMALL CELL LUNG CARCINOMA

96
Q

What is Imatinib used to treat?

A

CML caused by Philadelphia chromsome translocation

97
Q

What is Imatinib’s mechanism of action?

A

Small molecular inhibitor BCR-ABL constitutively active Tyrosine Kinase

98
Q

What are the mechanisms of resistance to Imatinib?

A

Point mutations in BCR-ABL that causes reduced drug affinity

99
Q

What is the mechanism of action of Asparaginase?

A
  • This is an enzyme used to treat childhood ALL
  • Hydrolyzes plasma L-asparagine into L-aspartate

Tumor cells cannot synthesize enough L-asparagine, but normal cells can; thus, this drug starves the tumor of essential nutrients

100
Q

What is the unique toxicity assocaited with Asaparaginase?

A

Anaphylaxis

101
Q

What is the mechanism of action of Bortezomib?

A

Proteasome inhibitor that elevates levels of p53 (important tumor suppressor)

p53 causes apoptosis in cancer cells*

102
Q

What is the major toxicity seen with Bortezomib?

A

Peripheral neuropathy

103
Q

What is the mechansim of action of Temsirolimus?

A
  • Inhibition of mTOR complex 1 (mTORC1)
  • mTORC1 is normally involved in protein translation
  • Thus, this drug it inhibits protein translation in rapidly dividing cells
104
Q

What are the unique side effects of Temsirolimus?

A

Hyperglycemia

Hypertriglyceridemia

105
Q

What is the resistance mechansim of Temsirolimus?

A
  • There are two mTORs, mTORC1 and mTORC2

- Temsirolimus inhibits mTORC1, which can cause an upregulation of mTORC2

106
Q

Which antineoplastic agents are nephrotoxic?

A

Cisplastin

Methotrexate

107
Q

Which antineoplastic agents are neurotoxic?

A
Vincristine 
Cytarabine
Cisplastin
Bortezomib 
Paclitaxel
108
Q

Which antineoplastic agents are cardiotoxic?

A

Doxorubicin

Trastuzumab

109
Q

Which antineoplastic agents cause bladder toxicity?

A

Cyclophosmphamide

110
Q

Which antineoplastic agents cause hypersensitivity reactions/ anaphylaxis?

A

Asparaginase

Paclitaxel

111
Q

What are 6-mercaptopurine and 6-thiguanine commonly used to treat?

A

These are purine analogs/ antimetabolites that inhibit s-phase; commonly used to treat AML and ALL

112
Q

What is Fludarabine commonly used to treat?

A

This is a purine analog/ antimetabolite that inhibits s-phase; it is commonly used to treat CLL

113
Q

What is Cladribine commonly used to treat?

A

This is a purine analog/ antimetabolite that inhibits s-phase; it is commonly used to treat Hairy Cell Leukemia